1
|
Cheney L, Barbaro JM, Berman JW. Antiretroviral Drugs Impact Autophagy with Toxic Outcomes. Cells 2021; 10:909. [PMID: 33920955 PMCID: PMC8071244 DOI: 10.3390/cells10040909] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 01/18/2023] Open
Abstract
Antiretroviral drugs have dramatically improved the morbidity and mortality of people living with HIV (PLWH). While current antiretroviral therapy (ART) regimens are generally well-tolerated, risks for side effects and toxicity remain as PLWH must take life-long medications. Antiretroviral drugs impact autophagy, an intracellular proteolytic process that eliminates debris and foreign material, provides nutrients for metabolism, and performs quality control to maintain cell homeostasis. Toxicity and adverse events associated with antiretrovirals may be due, in part, to their impacts on autophagy. A more complete understanding of the effects on autophagy is essential for developing antiretroviral drugs with decreased off target effects, meaning those unrelated to viral suppression, to minimize toxicity for PLWH. This review summarizes the findings and highlights the gaps in our knowledge of the impacts of antiretroviral drugs on autophagy.
Collapse
Affiliation(s)
- Laura Cheney
- Division of Infectious Diseases, Department of Medicine, Montefiore Medical Center and Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA
| | - John M. Barbaro
- Department of Pathology, Montefiore Medical Center and Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA; (J.M.B.); (J.W.B.)
| | - Joan W. Berman
- Department of Pathology, Montefiore Medical Center and Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA; (J.M.B.); (J.W.B.)
- Department of Microbiology and Immunology, Montefiore Medical Center and Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA
| |
Collapse
|
2
|
Su X, Weng S, Peng D. New Insights into Apolipoprotein A5 and the Modulation of Human Adipose-derived Mesenchymal Stem Cells Adipogenesis. Curr Mol Med 2020; 20:144-156. [PMID: 31560287 DOI: 10.2174/1566524019666190927155702] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 11/22/2022]
Abstract
Background:
The hallmark of obesity is the excessive accumulation of
triglyceride (TG) in adipose tissue. Apolipoprotein A5 (ApoA5) has been shown to
influence the prevalence and pathogenesis of obesity. However, the underlying
mechanisms remain to be clarified.
Methods:
Human adipose-derived mesenchymal stem cells (AMSCs) were treated with
600 ng/ml human recombinant ApoA5 protein. The effect of ApoA5 on intracellular TG
content and adipogenic related factors expression were determined. Furthermore, the
effect of ApoA5 on CIDE-C expression was also observed.
Results:
During the process of adipogenesis, ApoA5 treatment reduced the intracellular
accumulation of lipid droplets and the TG levels; meanwhile, ApoA5 down-regulated the
expression levels of adipogenic related factors, including CCAAT enhancer-binding
proteins α/β (C/EBPα/β), fatty acid synthetase (FAS), and fatty acid-binding protein 4
(FABP4). Furthermore, the suppression of adipogenesis by ApoA5 was mediated
through the inhibition of CIDE-C expression, an important factor which promotes the
process of adipogenesis. However, over-expressing intracellular CIDE-C could lead to
the loss-of-function of ApoA5 in inhibiting AMSCs adipogenesis.
Conclusions:
In conclusion, ApoA5 inhibits the adipogenic process of AMSCs through,
at least partly, down-regulating CIDE-C expression. The present study provides novel
mechanisms whereby ApoA5 prevents obesity via AMSCs in humans.
Collapse
Affiliation(s)
- Xin Su
- Department of Cardiovascular Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shuwei Weng
- Department of Cardiovascular Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Sangenito LS, Menna-Barreto RFS, d'Avila-Levy CM, Branquinha MH, Santos ALS. Repositioning of HIV Aspartyl Peptidase Inhibitors for Combating the Neglected Human Pathogen Trypanosoma cruzi. Curr Med Chem 2019; 26:6590-6613. [PMID: 31187704 DOI: 10.2174/0929867326666190610152934] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/11/2018] [Accepted: 08/23/2018] [Indexed: 12/11/2022]
Abstract
Chagas disease, caused by the flagellate parasite Trypanosoma cruzi, is a wellknown neglected tropical disease. This parasitic illness affects 6-7 million people and can lead to severe myocarditis and/or complications of the digestive tract. The changes in its epidemiology facilitate co-infection with the Human Immunodeficiency Virus (HIV), making even more difficult the diagnosis and prognosis. The parasitic infection is reactivated in T. cruzi/HIV co-infection, with the appearance of unusual manifestations in the chronic phase and the exacerbation of classical clinical signs. The therapeutic arsenal to treat Chagas disease, in all its clinical forms, is restricted basically to two drugs, benznidazole and nifurtimox. Both drugs are extremely toxic and the therapeutic efficacy is still unclear, making the clinical treatment a huge issue to be solved. Therefore, it seems obvious the necessity of new tangible approaches to combat this illness. In this sense, the repositioning of approved drugs appears as an interesting and viable strategy. The discovery of Human Immunodeficiency Virus Aspartyl Peptidase Inhibitors (HIV-PIs) represented a milestone in the treatment of Acquired Immune Deficiency Syndrome (AIDS) and, concomitantly, a marked reduction in both the incidence and prevalence of important bacterial, fungal and parasitic co-infections was clearly observed. Taking all these findings into consideration, the present review summarizes the promising and beneficial data concerning the effects of HIV-PIs on all the evolutionary forms of T. cruzi and in important steps of the parasite's life cycle, which highlight their possible application as alternative drugs to treat Chagas disease.
Collapse
Affiliation(s)
- Leandro S Sangenito
- Laboratorio de Estudos Avancados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Rubem F S Menna-Barreto
- Laboratorio de Biologia Celular, Instituto Oswaldo Cruz (IOC), Fundacao Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Cláudia M d'Avila-Levy
- Laboratorio de Estudos Integrados em Protozoologia, Instituto Oswaldo Cruz (IOC), Fundacao Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Marta H Branquinha
- Laboratorio de Estudos Avancados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - André L S Santos
- Laboratorio de Estudos Avancados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Ahmed D, Roy D, Cassol E. Examining Relationships between Metabolism and Persistent Inflammation in HIV Patients on Antiretroviral Therapy. Mediators Inflamm 2018; 2018:6238978. [PMID: 30363715 PMCID: PMC6181007 DOI: 10.1155/2018/6238978] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 09/06/2018] [Indexed: 12/30/2022] Open
Abstract
With the advent of antiretroviral therapy (ART), HIV-infected individuals are now living longer and healthier lives. However, ART does not completely restore health and treated individuals are experiencing increased rates of noncommunicable diseases such as dyslipidemia, insulin resistance, type 2 diabetes, cardiovascular disease, and nonalcoholic fatty liver disease. While it is well known that persistent immune activation and inflammation contribute to the development of these comorbid diseases, the mechanisms underlying this chronic activation remain incompletely understood. In this review, we will discuss emerging evidence that suggests that alterations in cellular metabolism may play a central role in driving this immune dysfunction in HIV patients on ART.
Collapse
Affiliation(s)
- Duale Ahmed
- Department of Biology, Carleton University, Ottawa, Ontario, Canada
| | - David Roy
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Edana Cassol
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
5
|
Sieprath T, Corne T, Robijns J, Koopman WJH, De Vos WH. Cellular Redox Profiling Using High-content Microscopy. J Vis Exp 2017. [PMID: 28570523 DOI: 10.3791/55449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Reactive oxygen species (ROS) regulate essential cellular processes including gene expression, migration, differentiation and proliferation. However, excessive ROS levels induce a state of oxidative stress, which is accompanied by irreversible oxidative damage to DNA, lipids and proteins. Thus, quantification of ROS provides a direct proxy for cellular health condition. Since mitochondria are among the major cellular sources and targets of ROS, joint analysis of mitochondrial function and ROS production in the same cells is crucial for better understanding the interconnection in pathophysiological conditions. Therefore, a high-content microscopy-based strategy was developed for simultaneous quantification of intracellular ROS levels, mitochondrial membrane potential (ΔΨm) and mitochondrial morphology. It is based on automated widefield fluorescence microscopy and image analysis of living adherent cells, grown in multi-well plates, and stained with the cell-permeable fluorescent reporter molecules CM-H2DCFDA (ROS) and TMRM (ΔΨm and mitochondrial morphology). In contrast with fluorimetry or flow-cytometry, this strategy allows quantification of subcellular parameters at the level of the individual cell with high spatiotemporal resolution, both before and after experimental stimulation. Importantly, the image-based nature of the method allows extracting morphological parameters in addition to signal intensities. The combined feature set is used for explorative and statistical multivariate data analysis to detect differences between subpopulations, cell types and/or treatments. Here, a detailed description of the assay is provided, along with an example experiment that proves its potential for unambiguous discrimination between cellular states after chemical perturbation.
Collapse
Affiliation(s)
- Tom Sieprath
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp; Cell Systems and Imaging Research Group (CSI), Department of Molecular Biotechnology, Ghent University
| | - Tobias Corne
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp; Cell Systems and Imaging Research Group (CSI), Department of Molecular Biotechnology, Ghent University
| | - Joke Robijns
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp
| | - Werner J H Koopman
- Department of Biochemistry , Radboud Institute for Molecular Life Sciences, Radboud University Medical Center
| | - Winnok H De Vos
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp; Cell Systems and Imaging Research Group (CSI), Department of Molecular Biotechnology, Ghent University;
| |
Collapse
|
6
|
Jensen K, Bikas A, Patel A, Kushchayeva Y, Costello J, McDaniel D, Burman K, Vasko V. Nelfinavir inhibits proliferation and induces DNA damage in thyroid cancer cells. Endocr Relat Cancer 2017; 24:147-156. [PMID: 28137980 DOI: 10.1530/erc-16-0568] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 01/30/2017] [Indexed: 12/20/2022]
Abstract
The HIV protease inhibitor Nelfinavir (NFV) inhibits PI3K/AKT and MAPK/ERK signaling pathways, emerging targets in thyroid cancers. We examined the effects of NFV on cancer cells that derived from follicular (FTC), papillary (PTC) and anaplastic (ATC) thyroid cancers. NFV (1-20 µM) was tested in FTC133, BCPAP and SW1736 cell lines. The effects of NFV on cell proliferation were determined in vitro using real-time microscopy and by flow cytometry. DNA damage, apoptotic cell death and expression of molecular markers of epithelial-mesenchymal transition (EMT) were determined by Western blot and real-time PCR. Real-time imaging demonstrated that NFV (10 µM) increased the time required for the cell passage through the phases of cell cycle and induced DNA fragmentation. Growth inhibitory effects of NFV were associated with the accumulation of cells in G0/G1 phase, downregulation of cyclin D1 and cyclin-dependent kinase 4 (CDK4). NFV also induced the expression of γH2AX and p53BP1 indicating DNA damage. Treatment with NFV (20 µM) resulted in caspase-3 cleavage in all examined cells. NFV (20 µM) decreased the levels of total and p-AKT in PTEN-deficient FTC133 cells. NFV had no significant effects on total ERK and p-ERK in BRAF-positive BCPAP and SW1736 cells. NFV had no effects on the expression of EMT markers (Twist, Vimentin, E- and N-Cadherin), but inhibited the migration and decreased the abilities of thyroid cancer cells to survive in non-adherent conditions. We conclude that NFV inhibits proliferation and induces DNA damage in thyroid cancer cell lines. Our in vitro data suggest that NFV has a potential to become a new thyroid cancer therapeutic agent.
Collapse
Affiliation(s)
- Kirk Jensen
- Department of PediatricsUniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Athanasios Bikas
- Department of Internal MedicineGeorgetown University Hospital MedStar, Washington Hospital Center Internal Medicine Residency Program, Washington, District of Columbia, USA
| | - Aneeta Patel
- Department of PediatricsUniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | | | - John Costello
- Department of PediatricsUniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Dennis McDaniel
- Uniformed Services University of the Health SciencesBiomedical Instrumentation Center, Bethesda, Maryland, USA
| | - Kenneth Burman
- MedStar Washington Hospital CenterEndocrinology, Washington, District of Columbia, USA
| | - Vasyl Vasko
- Department of PediatricsUniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
7
|
Erlandson KM, Lake JE. Fat Matters: Understanding the Role of Adipose Tissue in Health in HIV Infection. Curr HIV/AIDS Rep 2016; 13:20-30. [PMID: 26830284 DOI: 10.1007/s11904-016-0298-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
More than one-third of adults in the USA are obese and obesity-related disease accounts for some of the leading causes of preventable death. Mid-life obesity may be a strong predictor of physical function impairment later in life regardless of body mass index (BMI) in older age, highlighting the benefits of obesity prevention on health throughout the lifespan. Adipose tissue disturbances including lipodystrophy and obesity are prevalent in the setting of treated and untreated HIV infection. This article will review current knowledge on fat disturbances in HIV-infected persons, including therapeutic options and future directions.
Collapse
Affiliation(s)
- Kristine M Erlandson
- University of Colorado-Anschutz Medical Center, 12700 E 19th Ave, Mailstop B168, Aurora, CO, USA.
| | - Jordan E Lake
- University of California, Los Angeles, 11075 Santa Monica Blvd., Ste. 100, Los Angeles, CA, USA.
| |
Collapse
|
8
|
Sieprath T, Corne TDJ, Willems PHGM, Koopman WJH, De Vos WH. Integrated High-Content Quantification of Intracellular ROS Levels and Mitochondrial Morphofunction. ADVANCES IN ANATOMY EMBRYOLOGY AND CELL BIOLOGY 2016; 219:149-77. [PMID: 27207366 DOI: 10.1007/978-3-319-28549-8_6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oxidative stress arises from an imbalance between the production of reactive oxygen species (ROS) and their removal by cellular antioxidant systems. Especially under pathological conditions, mitochondria constitute a relevant source of cellular ROS. These organelles harbor the electron transport chain, bringing electrons in close vicinity to molecular oxygen. Although a full understanding is still lacking, intracellular ROS generation and mitochondrial function are also linked to changes in mitochondrial morphology. To study the intricate relationships between the different factors that govern cellular redox balance in living cells, we have developed a high-content microscopy-based strategy for simultaneous quantification of intracellular ROS levels and mitochondrial morphofunction. Here, we summarize the principles of intracellular ROS generation and removal, and we explain the major considerations for performing quantitative microscopy analyses of ROS and mitochondrial morphofunction in living cells. Next, we describe our workflow, and finally, we illustrate that a multiparametric readout enables the unambiguous classification of chemically perturbed cells as well as laminopathy patient cells.
Collapse
Affiliation(s)
- Tom Sieprath
- Cell Systems and Imaging Research Group (CSI), Department of Molecular Biotechnology, Ghent University, Ghent, Belgium.,Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Tobias D J Corne
- Cell Systems and Imaging Research Group (CSI), Department of Molecular Biotechnology, Ghent University, Ghent, Belgium.,Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Peter H G M Willems
- Department of Biochemistry (286), Radboud University Medical Centre (RUMC), Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Werner J H Koopman
- Department of Biochemistry (286), Radboud University Medical Centre (RUMC), Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Winnok H De Vos
- Cell Systems and Imaging Research Group (CSI), Department of Molecular Biotechnology, Ghent University, Ghent, Belgium. .,Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
9
|
Haser GC, Sumpio B. Systemic and cell-specific mechanisms of vasculopathy induced by human immunodeficiency virus and highly active antiretroviral therapy. J Vasc Surg 2016; 65:849-859. [PMID: 26994951 DOI: 10.1016/j.jvs.2016.01.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 01/15/2016] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Patients infected with human immunodeficiency virus (HIV) have higher rates of dyslipidemia, atherosclerosis, and chronic inflammation that can damage the vascular system compared with the general population. This can be attributed both to HIV itself and to highly active antiretroviral therapy (HAART) they receive. This review outlines the mechanisms by which HIV and HIV medications can cause vascular complications and identifies strategic areas of research to treat these dysfunctions. REVIEW HIV and HAART affect the vascular system through several mechanisms that target systemic or metabolic systems and specific cells. HIV causes dyslipidemia and chronic immune activation, which can contribute to atherosclerosis. In addition, HIV damages macrophages, endothelial cells, smooth muscle cells, and platelets, and this damage also plays a role in the development of atherosclerosis. HAART, particularly protease inhibitors, interferes with cholesterol metabolism and can affect macrophages, endothelial cells, and smooth muscle cells. The metabolic changes and cell damage induced by HIV and HAART put HIV patients at increased risk for atherosclerosis, dyslipidemia, and serious cardiovascular events such as myocardial infarction and stroke. CONCLUSIONS HIV patients have increased risk of developing potentially life-threatening cardiovascular pathology, which cannot be explained by traditional cardiovascular risk factors alone. More research is needed into therapies to target this HIV-specific vasculopathy.
Collapse
Affiliation(s)
- Grace C Haser
- Department of Vascular Surgery, Yale School of Medicine, New Haven, Conn
| | - Bauer Sumpio
- Department of Vascular Surgery, Yale School of Medicine, New Haven, Conn.
| |
Collapse
|
10
|
Foufelle F, Fromenty B. Role of endoplasmic reticulum stress in drug-induced toxicity. Pharmacol Res Perspect 2016; 4:e00211. [PMID: 26977301 PMCID: PMC4777263 DOI: 10.1002/prp2.211] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 12/14/2015] [Indexed: 12/13/2022] Open
Abstract
Drug‐induced toxicity is a key issue for public health because some side effects can be severe and life‐threatening. These adverse effects can also be a major concern for the pharmaceutical companies since significant toxicity can lead to the interruption of clinical trials, or the withdrawal of the incriminated drugs from the market. Recent studies suggested that endoplasmic reticulum (ER) stress could be an important event involved in drug liability, in addition to other key mechanisms such as mitochondrial dysfunction and oxidative stress. Indeed, drug‐induced ER stress could lead to several deleterious effects within cells and tissues including accumulation of lipids, cell death, cytolysis, and inflammation. After recalling important information regarding drug‐induced adverse reactions and ER stress in diverse pathophysiological situations, this review summarizes the main data pertaining to drug‐induced ER stress and its potential involvement in different adverse effects. Drugs presented in this review are for instance acetaminophen (APAP), arsenic trioxide and other anticancer drugs, diclofenac, and different antiretroviral compounds. We also included data on tunicamycin (an antibiotic not used in human medicine because of its toxicity) and thapsigargin (a toxic compound of the Mediterranean plant Thapsia garganica) since both molecules are commonly used as prototypical toxins to induce ER stress in cellular and animal models.
Collapse
|
11
|
Abstract
OBJECTIVE To review the mechanisms of anti-cancer activity of nelfinavir and other protease inhibitors (PIs) based on evidences reported in the published literature. METHODS We extensively reviewed the literature concerning nelfinavir (NFV) as an off target anti-cancer drug and other PIs. A classification of PIs based on anti-cancer mode of action was proposed. Controversies regarding nelfinavir mode of action were also addressed. CONCLUSIONS The two main mechanisms involved in anti-cancer activity are endoplasmic reticulum stress-unfolded protein response pathway and Akt inhibition. However there are many other effects, partially dependent and independent of those mentioned, that may be useful in cancer treatment, including MMP-9 and MMP-2 inhibition, down-regulation of CDK-2, VEGF, bFGF, NF-kB, STAT-3, HIF-1 alfa, IGF, EGFR, survivin, BCRP, androgen receptor, proteasome, fatty acid synthase (FAS), decrease in cellular ATP concentration and upregulation of TRAIL receptor DR5, Bax, increased radiosensitivity, and autophagy. The end result of all these effects is slower growth, decreased angiogenesis, decreased invasion and increased apoptosis, which means reduced proliferation and increased cancer cells death. PIs may be classified according to their anticancer activity at clinically achievable doses, in AKT inhibitors, ER stressors and Akt inhibitors/ER stressors. Beyond the phase I trials that have been recently completed, adequately powered and well-designed clinical trials are needed in the various cancer type settings, and specific trials where NFV is tested in association with other known anti-cancer pharmaceuticals should be sought, in order to find an appropriate place for NFV in cancer treatment. The analysis of controversies on the molecular mechanisms of NFV hints to the possibility that NFV works in a different way in tumor cells and in hepatocytes and adipocytes.
Collapse
Affiliation(s)
- Tomas Koltai
- Centro de Diagnostico y Tratamiento de la Obra Social del Personal de la Alimentación, Talar de Pacheco, Buenos Aires, 1618, Argentina
| |
Collapse
|
12
|
Zhu S, Cheng G, Zhu H, Guan G. A study of genes involved in adipocyte differentiation. J Pediatr Endocrinol Metab 2015; 28:93-9. [PMID: 25153216 DOI: 10.1515/jpem-2014-0002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Accepted: 06/20/2014] [Indexed: 11/15/2022]
Abstract
OBJECTIVE With the use of the microarray technique, genes expressed in the late phase of adipocyte differentiation were investigated. These genes play an important role in stimulating adipocyte growth and lipid droplet formation. Therefore, they contribute a great deal to the onset of obesity. METHODS With the use of SW872 adipocytes and the microarray technique, genes related to adipocyte differentiation were tested and compared with undifferentiated preadipocytes 14 days after induction. Real-time reverse transcription polymerase chain reaction (RT-PCR) was used for confirmation. RESULTS More than 21,329 transcriptors were expressed and determined, of which 1326 increased and 687 decreased undifferentiated adipocytes. Among them, 21 were highly expressed by more than 10-fold. With RT-PCR, 12 were confirmed, including apelin, CIDEC, PID1, LYRM1, ADD1, PPARγ2, ANGPTL4, ADIPOQ, ACOX1, FIP1L1, MAP3K2 and PEX14. Furthermore, genes involved in lipid metabolism, signal transduction, DNA replication, redox status and transcription factors were determined as well. Novel genes involved in adipogenesis (e.g., apelin) were detected. CONCLUSION A variety of genes were discovered and validated with RT-PCR at the late phase of adipocyte differentiation. This may help us better understand the onset of obesity and the potential role of adipocytes in other organs.
Collapse
|
13
|
Reyskens KMSE, Essop MF. HIV protease inhibitors and onset of cardiovascular diseases: a central role for oxidative stress and dysregulation of the ubiquitin-proteasome system. Biochim Biophys Acta Mol Basis Dis 2013; 1842:256-68. [PMID: 24275553 DOI: 10.1016/j.bbadis.2013.11.019] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/09/2013] [Accepted: 11/18/2013] [Indexed: 12/18/2022]
Abstract
The successful roll-out of highly active antiretroviral therapy (HAART) has extended life expectancy and enhanced the overall well-being of HIV-positive individuals. There are, however, increased concerns regarding HAART-mediated metabolic derangements and its potential risk for cardiovascular diseases (CVD) in the long-term. Here certain classes of antiretroviral drugs such as the HIV protease inhibitors (PIs) are strongly implicated in this process. This article largely focuses on the direct PI-linked development of cardio-metabolic complications, and reviews the inter-linked roles of oxidative stress and the ubiquitin-proteasome system (UPS) as key mediators driving this process. It is proposed that PIs trigger reactive oxygen species (ROS) production that leads to serious downstream consequences such as cell death, impaired mitochondrial function, and UPS dysregulation. Moreover, we advocate that HIV PIs may also directly lower myocardial UPS function. The attenuation of cardiac UPS can initiate transcriptional changes that contribute to perturbed lipid metabolism, thereby fueling a pro-atherogenic milieu. It may also directly alter ionic channels and interfere with electrical signaling in the myocardium. Therefore HIV PI-induced ROS together with a dysfunctional UPS elicit detrimental effects on the cardiovascular system that will eventually result in the onset of heart diseases. Thus while HIV PIs substantially improve life expectancy and quality of life in HIV-positive patients, its longer-term side-effects on the cardiovascular system should lead to a) greater clinical awareness regarding its benefit-harm paradigm, and b) the development and evaluation of novel co-treatment strategies.
Collapse
Affiliation(s)
- Kathleen M S E Reyskens
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa
| | - M Faadiel Essop
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa.
| |
Collapse
|